Angell 2016

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

www.MaterialsViews.

com

DNA Nanotechnology

DNA Nanotechnology for Precise Control over Drug


Delivery and Gene Therapy
Chava Angell, Sibai Xie, Liangfang Zhang, and Yi Chen*

From the Contents Nanomedicine has been growing exponentially due to its
enhanced drug targeting and reduced drug toxicity. It uses
1. Introduction ........................................ 1118 the interactions where nanotechnological components
2. DNA Nanotechnology ........................... 1118 and biological systems communicate with each other
to facilitate the delivery performance. At this scale, the
3. DNA-Based Approaches for physiochemical properties of delivery systems strongly
Drug/Gene Delivery ............................. 1120 affect their capacities. Among current delivery systems,
4. Summary and Future Outlook............... 1129 DNA nanotechnology shows many advantages because
of its unprecedented engineering abilities. Through
molecular recognition, DNA nanotechnology can
be used to construct a variety of nanostructures with
precisely controllable size, shape, and surface chemistry,
which can be appreciated in the delivery process. In this
review, different approaches that are currently used for
the construction of DNA nanostructures are reported.
Further, the utilization of these DNA nanostructures with
the well-defined parameters for the precise control in drug
delivery and gene therapy is discussed.

small 2016, 12, No. 9, 1117–1132 © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.small-journal.com 1117
reviews
www.MaterialsViews.com

1. Introduction 2. DNA Nanotechnology


Naked therapeutic agents, which include small-molecule and DNA Nanotechnology is a branch of nanoengineering that
biomolecular drugs, have some intrinsic issues that prevent utilizes the precise and predictable nature of complemen-
the full realization of their functions in the body. This includes tary DNA base pairing to create 2D and 3D structures. This
poor solubility, poor stability against chemical and enzymatic branch of nanotechnology has many applications in medicine
degradation, inability to pass the biologic barriers, unwanted and biotechnology as DNA can easily be functionalized with
side effects and toxicity. To overcome these challenges, many binding ligands and fluorescent dyes for signal readout.[13]
different drug delivery systems have been developed in the The field was pioneered in the 80’s by Ned Seeman, who pro-
past decades.[1,2] They can be generally classified into two posed the construction of an immobile 4-way arm junction
categories: natural and biomimetic delivery systems such comprised of DNA and the connecting networks through
as virus[3] and red blood cells mimicking carriers,[4] and sticky ends.[14]
synthetic delivery systems such as liposomes and polymeric After a few decades of development, this field has been
particles.[5,6] These delivery platforms use natural or synthetic exponentially explored. Varies of DNA nanostructures and
particles to carry drugs and thus bypass their physiochem- applications have been successfully reported. Recently, DNA
ical limitation and barriers for effective delivery. Through nanostructures start to be used as a creative solution to both
appropriate parameter design, these delivery systems can the accumulation and off target effects of other drug delivery
effectively increase the drug loading efficacy, the circulation modalities as it is a nontoxic, biologically derived material
time in body, and improve the final therapeutic results.[7,8] that can be easily functionalized to target receptors on the
Many of them have entered clinical trials and some have cell surface and can also be covalently or electrostatically
been approved for clinical use. attached to metallic nanostructures.[15]
Although these drug carriers are actively being To create stable structures out of DNA, certain sequence
developed, there is still a measurable gap that has to be symmetry has to be minimized to prevent the instability that
surmounted to achieve the ultimate goal of drug delivery: is seen in many intermediate structures, such as Holliday
the maximal therapeutic efficacy with minimal toxic effects. junctions (Figure 1A).[14] There are also many design consid-
In spite of different routes of administration, including erations such as the incorporation of sticky ends, and recip-
oral, injection, transdermal and inhalation, the delivery rocal exchanges for stability, also known as ‘crossovers’.[16]
systems and the therapeutic cargos can be traced to the cel- DNA nanotechnology is a highly intriguing strategy for
lular level. This stimulates the exponential development of self-assembled drug delivery structures as it allows for the
nanomedicine which uses nanotechnology tools to enhance creation of precise structures that can mimic nanoparticles or
drug targeting, reduce drug toxicity, and enable reliable viral capsids. Such structures include polyhedra of different
diagnosis.[9,10] Among these newly developed delivery sys- sizes and shapes such as cubes,[17] tetrahedra,[18–20] octa-
tems, DNA nanostructures show high promise. DNA is hedra,[21] buckyballs,[19] and icosahedra.[22,23] Larger shapes
a biopolymer that can self-assemble into double helices based on the DNA origami method are also reported.[24]
through by Watson–Crick base pairing and is stabilized by Goodman et al. reported a facile way of building DNA tet-
hydrogen bonds, p–p stacking, and hydrophobic interac- rahedra using four different strands, with the struts less than
tions. They have well-defined structures: the helical turn is 10 nm in length.[18] Under the simple process of cooling from
3.4 nm and the diameter is ≈2.2 nm for the B-form double- 95 °C to 4 °C in 30 s, tetrahedra form under a yield of 95% and
helical molecules. Fueled by this base-pairing ability, DNA can be characterized by a single band on a native gel.
nanotechnology has enabled many strategies to form DNA Another approach was proposed by He et al. to construct
nanostructures with well-controlled size, shape, and surface tetrahedra, dodecahedra, and bucky ball structures through
chemistry. In nanomedicine, these properties strongly affect DNA tiles.[19] The proposed three-point star motif was used
the delivery capacities.[11,12] and thus give DNA nanostruc- for 2D crystal formation.[25] In this approach, the flexibility
tures unique control abilities to address some of the unmet of the long central strand was enhanced by elongating the
delivery requirements. non-binding hinge parts which they call “loops”, so that the
This review will discuss the applications and weigh the motifs tend to bend and form closed structures. The concen-
usefulness and viability of DNA nanostructures in precision tration of the motif also affects the resulting structure. A loop
nanomedicine. The discussion will begin with an introduc- length of 5 bases and a low concentration of 75 nm gives a
tion to DNA nanotechnology, including some of the types of final result of tetrahedra, while 3 bases loops and 50 nm gives
structures that can be formed. Following this will be an elu- dodecahedra, and 3 bases loops and 500 nm concentration
cidation of the DNA oligonucleotide approach, the original gives buckyball structure. Each strut is around 14 nm.
strategy used to create DNA nanostructures, and current uses
of this strategy for drug delivery. The next motif that will be
discussed is the DNA origami strategy and what has currently C. Angell, S. Xie, Prof. L. Zhang, Prof. Y. Chen
been achieved with DNA origami in drug and gene therapy Department of NanoEngineering
as well. Finally, we will touch on some of the unique shapes University of California
San Diego, La Jolla, CA 92093, USA
and modalities that have been created for the purpose of
E-mail: yic047@ucsd.edu
DNA drug delivery and finish with our remarks and thoughts
on the future viability of this field. DOI: 10.1002/smll.201502167

1118 www.small-journal.com © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim small 2016, 12, No. 9, 1117–1132
www.MaterialsViews.com

Figure 1. Demonstration of DNA nanostructures. A) immobile 4-strand junction proposed by Seeman. Reproduced with permission.[14] Copyright
1983, Cell Press. B) Icosahedral structure formed by flexible five-point-star motifs. Reproduced with permission.[22] Copyright 2008, National
Academy of Sciences. C) Truncated cubic structure. Reproduced with permission.[17] Copyright 1991, Nature Publishing Group. D) Different
structure formed by the same three-point-star motif with different “loop” length and concentration. Reproduced with permission.[23] Copyright
2009, Wiley-VCH.[23] E) Single-stranded DNA bricks form 3D canvas. Reproduced with permission.[27] Copyright 2012, American Association for the
Advancement of Science (AAAS).

Similar tile approach can also be used to form icosahe- The icosahedra sample was prepared in the concentration of
dral DNA nanostructure (Figure 1B).[22] The same group 20 nm to prevent the formation of larger structure. DNA
reported in 2008 a method to fabricate truncated icosahedra nanostructures can also be formed through step-by-step pro-
of diameter ≈20 nm. The five point star motif used in this cess. Chen et al. demonstrated this method by constructing a
method has a four base loop. They also reported that the nano-scale cube structure (Figure 1C).[17] In each step they
concentration of the motif also has an effect on the predomi- hybridize two squares and in the last step the ends are con-
nant structure that can form: the samples form a big cage nected to form a cube. Bahtia et al. reported a three-step
structure (≈115 nm in diameter) under higher concentration. way to encapsulate gold nanoparticles into a truncated DNA

small 2016, 12, No. 9, 1117–1132 © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.small-journal.com 1119
reviews
www.MaterialsViews.com

nanostructure (Figure 1D).[23] They first anneal DNA strands


to form five-arm vertex motifs, and then combine the two
“halves” form icosahedron with complementary binding sites.
The two halves were put into a gold nanoparticle solution.
The icosahedron with diameter of roughly 19 nm can success-
fully encapsulate 2–3 nm diameter gold nanoparticles. Other
icosahedra have been reported that can intercalate doxoru-
bicin (Dox).[26]
An important concept in DNA nanotechnology is ease
of modular self-assembly. In 2012, Ke et al. reported the con-
struction of short “DNA bricks” that can be used to create
many different types of structures without having to redesign
staple strands and scaffolding routing as is necessary in cre- Figure 2. Intercalation of Doxorubicin. Reproduced with permission.[31]
ating varying origami structures (Figure 1E).[27] The authors Copyright 2014, Elsevier.
reported essentially a lego-like modality in which each brick
had a distinct shape and could bind to 4 neighbors just as a the introduction of the artificial siRNA can monopolize the
lego can fit into specific holes. These interactions are defined natural interference pathway of the RNA silencing complex,
by the domains of the DNA bricks which have unique which is an important part of the cellular function.[35] One
sequences. Therefore, each 32 nucleotide brick is distinctive of the biggest challenges in RNA delivery is stability in vivo,
and can be combined to create different 3D structures with but chemical modifications of the oligomers can increase the
both controllable surface features and precise cavities within stability.[36]
the structure. 102 different structures were created using this The targeting issue using DNA nanostructures can be
method merely by selecting the bricks needed which greatly achieved through the use of DNA aptamer sequences and
simplifies the process of creating 3D DNA nanostructures.[27] also through the use of functionalizing the DNA with tar-
There are several methods to make viable, stable struc- geting ligands and can help avoid many of the undesirable off
tures comprised mostly or entirely of DNA: DNA origami, target effects.[32,37,38] For example, Zhu et al. created synthetic
oligonucleotide design, and rolling circle amplification. In DNA adducts to allow for targeted anticancer delivery.[32]
this review, we will touch on the methodologies used to These take advantage of the overexpression of certain recep-
form DNA nanostructures and how those methodologies are tors during cancer.[39]
being coupled with both gene silencing therapies and cancer Once been uptake by cells, DNA nanostructure can be
therapeutic drugs to more successfully deliver and control designed to respond to environmental triggers to release
the delivery of therapeutic modalities to site specific cells loaded drugs, such as incorporation of linkers that are
in vivo. responsible to changes in the pH.[29,30,40,41] This technique can
also aid in mapping the pH changes inside the cells itself, as
reported by Modi et al.[41]
3. DNA-Based Approaches for Drug/Gene There are several possible pathways for these DNA
Delivery nanostructures to enter the cell and partially be explored by
using inhibitors to block certain pathways.[42,43] These path-
There are many methods that have been coupled to DNA ways can be divided into two types: endocytotic pathways
nanotechnology to effectively deliver therapeutic modalities, and non-endocytotic pathways.[44] The endocytotic pathways
including drug modalities, gene-silencing modalities, and the include phagocytosis, clathrin mediated endocytosis, caveolae
incorporation of cellular targeting into DNA nanostructures. mediated endocytosis, and macropinocytosis. However, it is
Many DNA drug delivery systems include the incorporation not well understood what mechanism is responsible for the
of doxorubicin (Dox), an anticancer therapeutic,[28,29] as the successful uptake of DNA nanostructures. Preliminary data
proof of concept for small molecular drug delivery. Dox can such as that demonstrated by Chen et al. suggested clathrin
intercalate into the DNA structure and allow it to form prop- receptor mediated endocytosis.[43] They measured the mecha-
erly by relieving torsional stress, as showed in Figure 2.[29–31] nism of the uptake of their rolling circle amplification (RCA)
However, despite Dox’s potency as an anticancer drug, it nanoribbons through inhibitors such as chlorpromazine, a
can have serious side effects when it accumulates off-target clathrin inhibitor, genistein, a caveolae mediated inhibitor,
such as cardiomyopathy, which can lead to congestive heart methyl-β-cyclodextrin, a lipid-raft endcytosis inhibitor, wort-
failure.[32] Therefore, avoiding off-target or premature mannin, a macropinocytosis inhibitor, and NaN3, an energy
delivery must be achieved. dependent endocytosis inhibitor. Final results suggested
Another therapeutic technique that many researchers that the uptake is clathrin mediated and lipid raft mediated
have been taking advantage of is gene-silencing through RNA endocytosis.
interference.[33] Small interfering RNA (siRNA) in particular Chen et al. also studied the mechanism of transfection of
has been utilized by many researchers for therapeutic gene DNA nanoparticle (DNPs) and suggested that nucleolin is a
silencing and can be modified to be target specific to avoid good target for non-viral gene delivery with the presence of
off target silencing effects.[34] RNA interference can usually glucocorticoid receptor (GCR).[45] Besides the disruption of
be used without triggering the immune system. However, a raft protein, flotillin also decrease the transfection of DNPs,

1120 www.small-journal.com © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim small 2016, 12, No. 9, 1117–1132
www.MaterialsViews.com

while inhibition of other endocytic pathways barely affects into the structure without affecting the shape. The Mucin-1
the process. (MUC-1) aptamer was used to target MUC 1+ MCF-7 cells.
Most of the cell membranes carry net negative charge The dox–apt–icosahedron complex shows great specificity
and nucleic acids also carry a net negative charge.[46] As a and aptamer-mediated internalization for killing epithelial
result, some DNA nanostructures, not containing targeting cancer cells.
modalities, may need some kind of cationic transfection DNA polyhedral nanostructures have also been used in
agent, such as Lipofectamine, to increase the cellular uptake gene delivery. Lee et al. reported a way to deliver siRNA
and therapeutic efficacy.[46] Positively charged intercalators using a tetrahedral DNA structure with a hydrodynamic size
or positively charged proteins, such as Dox, viral capsule of 28.6 nm with narrow distribution (Figure 3C).[52] Molec-
proteins, or cationic polymers, may also help reduce the net ular identical oligonucleotide particles (ONPs) with six folic
negative charge thus reducing the electrostatic repulsion in acid ligands per particle were injected in nude mice with
the DNA structure itself and between the DNA nanotech- human epidermoid carcinoma cell (KB) xenograft tumors.
nology and the cell membrane.[33,47–49] Results show that the ODNs can accumulate primarily in
For the purpose of this review, we will discuss the the tumor and kidney. The circulation time of the ODNs
different mechanisms of DNA nanostructure formation as is also a lot longer than the parent siRNA (blood half-life
separate categories. These mechanisms are integral to what t1/2 ≈ 24.2 min vs 6 min).
can be achieved for the shape and size of the structures and A concept of nanotrain was reported by Zhu et al. in
can affect what can be delivered and the release kinetics of 2013.[53] Two strands of ODNs alternatively bind to each
delivery. Some large structures can only be created through other triggered by an aptamer trigger to form a longer chain
the use of DNA origami and certain modularity can only be train-like structure with an aptamer sgc8 on one end to target
achieved using the oligonucleotide approach. However, some specific cells (Figure 3D). Dox is intercalated into the DNA
structures are hybrids of different fabrication methods and double strand. The complex can be specifically transport the
may fit multiple synthesis categories. Therefore, they will be drug to target cancer cells, initiate endocytosis and unload
categorized by their primary method of synthesis. the drugs within the cells. The complex was tested in vivo
in NOD.Cg-Prkdc (scid) IL2 mice and showed reduced side
effects and stronger therapeutic potency.
3.1. Oligonucleotide Approach Wu et al. reported a modular aptamer based DNA
nanoassembly (AptNA) approach (Figure 3E).[54]
Due to the precise and predictable pairing nature of DNA Multifunctional DNA sequences are first assembled into
chains, the size and shape of the nanoscale DNA constructs Y-shaped functional domains and then connected to
can be designed to fit current drug delivery needs. Moreover, X shaped connectors. The connector has a photopolymeriz-
overhangs can be introduced on to the structures to allow able arm so that the building unit with different functional
easy functionalization of fluorescent dyes, targeting agents or domains could be polymerized into spherical structure of
even drugs. diameter around 200 nm. The aptamer sgc8 was used to
The oligonucleotide strategy was reported first by target CCRF-CEM (T cell acute lymphoblastic leukemia
Nadrian Seeman in 1983 and is widely considered the foun- cell line) cancer cells. The cells showed increased viability
dation of DNA nanotechnology. He synthesized a stable compared to the treatment by free Dox. The DNA nanopar-
4 arm junction with 4 sixteen base strands by minimizing the ticles also showed selectivity on CEM cells rather than the
strand symmetry.[14] The oligonucleotide strategy requires non-target Ramos cells.
more design consideration as there are no viable programs Li et al. developed a tetrahedron motif bearing unmethyl-
for designing these structures. Oligonucleotides have been ated cytosine–phosphate–guanine (CpG) motifs which have
used to create many types of structures such as nanotubes,[50] immunostimulatory properties.[55] This is due to the fact that
tetrahedra,[13,51] icosahedra,[23] and many other polyhedral these motifs can bind to certain cellular receptors known as
structures.[19,21] endosomal Toll-like receptor 9 and trigger an immunostim-
In 2010 Kim et al. reported a simple method of inter- ulatory response. After incubation with macrophage cell
calating Dox into DNA tetrahedra as carriers to reverse line (RAW264.7 cells), these structures were mostly local-
drug resilience of cancer cells.[13] The dye Cy5 was incor- ized in the cytoplasm as imaged by confocal microscopy.
porated into the tetrahedron for characterization purpose Degradation studies were also conducted by incubating the
(Figure 3B). Results show that even without any targeting tetrahedrons in fetal bovine serum (FBS). These structures
agent, the DNA tetrahedra are able to enhance uptake and remained stable at 4 hours whilst simple DNA duplexes
bypass the efflux process in multidrug resistant breast cancer degraded in the span of 2 hours.
cell line (MCF-7 cancer cells). These results suggest certain Sellner et al. reported in 2015 an in vivo study of gene
shape dependence on transfection efficacy. delivery by DNA nanotubes. The nanotubes were assem-
An example of DNA polyhedra assembled from oligo- bled by the single stranded tile (SST) method, in which each
nucleotides was reported by Chang et al. in 2011.[26] They tile consists of four 10–11 bases domains and a total length
designed a six-point-star motif with five sticky ends that are of 42 bases.[50] Sticky ends of poly-As stick out of the tube
self-complementary and one end with targeting aptamer to side walls for functionalization. CpG and fluorescent dyes
assemble into icosahedra with diameter of around 28 nm, ATTO488-dUTP or Cy3-dUTP were conjugated onto the
while the ends with aptamer erect. Dox was intercalated structure. The in vivo study on anesthetized mice showed

small 2016, 12, No. 9, 1117–1132 © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.small-journal.com 1121
reviews
www.MaterialsViews.com

Figure 3. A) The original DNA tetrahedral structure proposed by Turberfield. Reproduced with permission.[18] Copyright 2005, AAAS. B) Modified
DNA tetrahedral structure with Cy5 dye and Dox intercalation. Reproduced with permission.[13] Copyright 2013, Royal Society of Chemistry.
C) Incoporation of siRNA onto the struts of the DNA nanotetrahedron. Reproduced with permission.[52] Copyright 2012, Nature Publishing Group.
D) Proposed drug delivery route of DNA nanotrain.[53] E) Multifunctional aptamer-based nanoparticles. Reproduced with permission.[54] Copyright
2013, American Chemical Society.

that while plain nanotubes do not induce an immune than that, other strategies for synthesizing precision DNA
response, CpG conjugated nanotubes can target tissue-res- nanotechnology are also extremely attractive for their shape
ident macrophages and reach intact muscle tissue almost control, control of flexibility and size, and ability to create
immediately, and elevate immune response strongly. The larger, modular structures that can even be extended to
DNA nanotubes also had some stability against DNase I include kinematic, movable joints.[27,56,57]
within muscle tissue. This approach is normally called DNA origami. It was
pioneered by Paul Rothemund in 2006 (Figure 4B).[58] DNA
origami refers to the folding of a long stranded bacteriophage
3.2. Origami Approach (7240 nt) through the use of over 200 complementary staple
strands to fold the backbone. Paul Rothemund used the
As seen above, DNA oligonucleotides allow for the crea- M13m18 bacteriophage and his original structures were 2D.
tion of precise structures that can be manipulated to deliver Each oligonucleotide is considered a 6 nm pixel to build any
both gene therapies and anticancer therapeutics. Other pattern of desire. By putting sticky ends on the connection

1122 www.small-journal.com © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim small 2016, 12, No. 9, 1117–1132
www.MaterialsViews.com

Figure 4. A) DNA octahedron obtained by folding a 1.7 kilobase backbone. Reproduced with permission.[63] Copyright 2004, Nature Publishing
Group. B) Rothemund’s DNA origami shapes. Reproduced with permission.[58] Copyright 2006, Nature Publishing Group. C) 3D DNA origami
structures. Reproduced with permission.[60] Copyright 2009, Nature Publishing Group. D) Large 3D structures synthesized by Ilumina to carry
therapeutic loads. Reproduced with permission.[24] Copyright 2014, AAAS.

part of each construct, an extended array can also be formed. modality and facilitating its use for structural formation and
Since then, the DNA origami motif has been extended to drug delivery.
three dimensions.[59,60] There are now several programs Shih et al. reported back in 2004 an idea similar to the
that facilitate the design of DNA origami structures such as later developed DNA approach, which is to fold a long
caDNAno and SARSE.[56,61] With DNA origami, structures backbone DNA chain by binding it at different points using
such as smiley faces, tetrahedrons, DNA nanotubes, DNA staple strands (Figure 4A).[63] In this example a 1669-nucle-
barrels, DNA “dolphins” and many other shapes are pos- otide single-stranded DNA is used as the backbone and
sible.[20,28,56,58,62] Many groups have worked on extending this five 40-nucleotide strands are used to fold the backbone.

small 2016, 12, No. 9, 1117–1132 © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.small-journal.com 1123
reviews
www.MaterialsViews.com

Double-crossover and paranemic-crossover structures are compared them to structures with a natural twist density of
designed to ensure the stiffness of the structure. It was 10.5 base pairs per turn. The increased binding affinity of the
proposed that the long backbone chain can be readily Dox also translates into both a higher loading and a slower
amplified. and more sustainable release profile.[28]
Douglas et al. reported that using the same DNA ori- Other groups have also reported utilizing DNA origami
gami method, it is also possible to fabricate 3D structures.[61] structure for Dox intercalation and controlled delivery.
They used honeycomb-pleat-based strategy to bring the Zhang et al. reported the construction of varying shapes
helices together at a certain angle to construct 3D struc- composed of DNA origami structures and loaded these
tures (Figure 4C). They also developed a software called structures with Dox to measure the in vitro drug release
caDNAno, an easy tool to design 3D structures and program and the in vivo antitumor effects (Figure 5B).[62] The shapes
the staple strands. The whole design and synthesis process reported were a triangle, a square, and a tube. To test the
took 2 weeks as they reported. accumulation, each structure was loaded with quantum dots
One of the largest challenges that faced DNA origami and imaged fluorescently. The triangle shape demonstrated
was the time it takes to fold and hybridize the structures. In the highest efficacy of passive tumor targeting out of the
2012, Sobczak et al. determined that a long annealing pro- three structures and maintained accumulation levels for 24
cess is not necessary to create these structures in a “one-pot” hours. Both the square and tube showed higher levels of
reaction scheme as there is a distinct range where DNA will off-target accumulation in the liver and kidney. The struc-
fold and hybridize.[64] These experiments also reported that tures were stable in serum for 24 hours and demonstrated
DNA folding and unfolding are non-equilibrium processes as slow release properties with 20% released after 8 hours. In
the temperature for unfolding is higher than the one at which more acidic buffer conditions, which more closely mimic a
the structures formed. This experiment allows DNA origami tumor microenvironment, the release content was increased.
to be on a more feasible time scale. When tumor-bearing mice were injected with saline, Dox,
There is the desire within this field to create large struc- Dox/origami structures, and origami structures, the Dox/
tures that mimic viral capsids. Iinuma et al. reported the con- origami structures were the most effective at reducing the
struction of polyhedra from million daltons (MD) monomers tumor size. Earlier experiments reported by this group dem-
(Figure 4D).[24] These monomers created a 5-MD DNA ori- onstrated that these nanostructures helped avoid multidrug
gami three arm junction that was sixty times more massive resistance that Dox can induce by testing the Dox/origami
than previously reported 3-arm motifs. These junctions have structures in MCF 7 Dox resistant cells which were created
a “dynamic connector” design and Iinuma et al. created a by exposing the cells to increasing concentrations of Dox
DNA tetrahedron, a cube, a triangular prism, a pentagonal (Figure 5C).[66]
prism, and a hexagonal prism. All structures were on the As mentioned earlier during the course of this review,
order of 20–60 MD and had compartments that are similar effective transfection and uptake of DNA nanostructures can
in size to those of bacterial microcompartments and could be be difficult and lead to low yield and therapeutic dose due to
used to carry therapeutic loads. their net negative charge. Mikkilä et al. coated DNA origami
In drug delivery, the kinetics and controllability of structures with cowpea chlorotic mottle virus capsid proteins
release is an important factor for consideration. If the thera- to allow for a more effective transfection as the proteins have
peutic is released in a burst mechanism as opposed to a slow, a positively charge N-terminus.[48] This helps to combat the
tunable release, there will be a higher initial delivery, how- net negative charge of the DNA and allowed the structures
ever this can lead to high physiological damage and a lower to enter the cell. The DNA origami nanostructures “tem-
lifetime of the therapy, which leads to the need for higher plate” the self-assembly of the virus proteins with high yield
dosages.[65] DNA origami allows for either controlled or trig- through electrostatic binding. The results reported transfec-
gered release of a therapeutic modality through either the tion efficiency higher than Lipofectamine 2000. The proposed
intercalation of positively charged molecules or the linking uses for these structures are drug delivery applications.
of certain peptides or proteins onto the surface of the DNA Another method for utilizing DNA origami for delivery is
origami structure. covalently linking the small molecule drugs or nanoparticles
In 2012, Zhao et al. designed and fabricated a structure to the structure. In 2012, Douglas et al. reported the construc-
that has controllable release properties of Dox, a well-known tion of logic gated nanorobot with a hexagonal barrel shape
anticancer therapeutic. These structures take advantage of (Figure 5D).[67] This barrel can be loaded with therapeutics
the fact that DNA origami structures with a specific twist through the use of covalent linkers on the inside. The logic gate,
density will only correctly fold in the presence of an interca- or “lock”, is a 23 base pair sequence that is antigen activated.
lating molecule such as Dox (Figure 5A).[28] This was origi- This type of sequence is known as an aptamer and its sensitivity
nally demonstrated by Ke et al. who inserted base pairs (bp) is directly related to the length of the sequence. In the pres-
in between crossover junctions.[47] These insertions, while ence of the correct antigen, the duplex will bind to the antigen
relieving electrostatic repulsion, also increase torsional strain, and allow the barrel to open. The length of the sequence is
thus destabilizing the structure and allowing for the inter- tuned such that the risk of spontaneous activation is minimized
calations to bind to the structure.[47] This binding relieves (at around 16 bp) whilst the sensitivity is preserved as longer
the stress and encourages the correct folding of the origami sequences are much less sensitive. These aptamer-based cues
structures. Zhao et al. created structures with 12 base pairs allow for triggering and targeted delivery based on local envi-
per turn that only fold correctly in the presence of Dox and ronments that are specific to certain disease modalities.

1124 www.small-journal.com © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim small 2016, 12, No. 9, 1117–1132
www.MaterialsViews.com

Figure 5. A) Doxorubicin-intercalating nanotubes. Reproduced with permission.[28] Copyright 2012, American Chemical Society. B) Doxorubicin-
intercalating nanostructures. Reproduced with permission.[62] Copyright 2014, American Chemical Society. C) Doxorubicin-intercalating
nanostructures in combating drug resistance. Reproduced with permission.[66] Copyright 2012, American Chemical Society. D) DNA origami
nanorobot. Reproduced with permission.[67] Copyright 2012, AAAS.

3.3. Rolling Circle Approach polymerase continually adds complementary nucleotides to the
template until it is denatured or runs out of available nucleo-
Rolling circle amplification (RCA) represents a novel way tides. This creates long stranded DNA with a known, repeating
to approach precision DNA nanostructure construction. sequence that is dependent on the sequence of the circular
RCA creates long stranded structures with repeating DNA template.[49] This long stranded DNA can be manipulated
sequences through the use of a circular template and DNA through staple strands similarly to DNA origami.[43,69] The
polymerase, which copies the template thousands of times.[68] staple strands can also be manufactured through RCA and
This allows for the precise templating of the RCA strand. The cleaved through a site-specific cleavage. RCA has been used

small 2016, 12, No. 9, 1117–1132 © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.small-journal.com 1125
reviews
www.MaterialsViews.com

Figure 6. A) DNA origami using rolling circle amplification. Reproduced with permission.[69] Copyright 2013, Wiley-VCH. B) RNAi microsponge.
Reproduced with permission.[73] Copyright 2012, Nature Publishing Group. C) DNA nanobelts loaded with reporter/therapeutic molecules and cell
penetrating peptides. Reproduced with permission.[71] Copyright 2015, Wiley-VCH.

to create nanotubes,[70] DNA nanoballs.[49] nanobelts that can DNA (Figure 6A).[69] These structures can easily be folded
intercalate dox and quantum dots,[71] and DNA nanoclews.[29] by fewer staple strands than DNA origami and can be uti-
It has also been incorporated into DNA logic gates.[72] lized for therapeutic, drug delivery purposes as they are
RCA can also be applied similarly to DNA origami, as easily internalized by cells.[43,69]
demonstrated by Ouyang et al. who created a long stranded Yan et al. has demonstrated a method to integrate nano-
DNA template out of RCA instead of using a bacteriophage particles and DNA strands produced in rolling circle

1126 www.small-journal.com © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim small 2016, 12, No. 9, 1117–1132
www.MaterialsViews.com

amplification (RCA) utilizing a similar method to Ouyang creation of unique structures with repeating units. These
(Figure 6C).[71] DNAs are grown in-situ on gold nanopar- repeating units are critical in allowing the precise control of
ticle surface, and the repetitive long chains are folded into the nanostructure degradation, through the incorporation of
so-called DNA belts while remaining at the particle surface. regions with known responses to cellular environments, and
Drugs and targeting agents can be loaded on to the DNAs therefore, therapeutic delivery.
by either merging or intercalating mechanism for theranostic
purposes. Merging was used to load quantum dots (QDs)
for cellular imaging and intercalation was used to load Dox. 3.4. DNA Nanoparticles
Incorporation of the QDs and cellular penetration peptides
(CPPs) on the nanoparticle (NP)–DNA conjugate gives QDs Another possible modality for utilizing DNA as a precision
the ability to get into U87 MG cells, while maintaining the drug or gene delivery vehicle is to create nanoparticles
spacing so that QDs are not quenched by CPPs. Incorpora- comprised of the DNA itself (Figure 7), or a DNA
tion of Dox into the structure also shows a 2.5 times higher nanoparticle.[45,74–89] DNA nanoparticles feature controllable
cell-killing efficacy in U87 MG cells than pure Dox. sizes from ≈20 nm to ≈200 nm in diameter. The fabrication of
Another proposed use for RCA is the self-degradable the particles also allows a polyethylene glycol shell for longer
nanoclew reported by Sun et al.[29] The nanoclew is woven circulation time.[80–84] It is also proved facile to functionalize
by RCA and includes DNase to degrade and release a the particle with targeting groups like serpin enzyme complex
therapeutic load inside the cells. To enhance the loading of receptor, SEC.[76]
the anticancer therapeutic Dox, multiple G–C sequences In this approach, DNA nanoparticles that were trans-
were incorporated. The nanoclew is a composite structure formed from complexes of DNA to nanoparticles from
including a polymeric capsule which contains the DNase and 18–25 nm through the use of poly-l-lysine covalently
is degradable by acidic pH, allowing for triggered release by coupled to a synthetic peptide.[74] Longer chain length
the cellular environment (i.e., the acidity of the late stage (53.7 kDa poly-K) gives smaller particles and significantly
endosome) and protecting the Dox until it can have the most more and longer duration of gene expression than short
therapeutic effect. The capsule is positively charged to allow chain (9.7 kDa poly-K) complex. Moreover, shorter peptide
for incorporation into the structure. Finally, these structures chains give less protection from the DNase both in vitro and
were incorporated with folic acid to allow for the targeting in vivo.[77]
of the folate receptors which are overexpressed in many can- This complex allows the gene transfer into the cell via cer-
cers. These structures showed both efficient internalization tain cellular receptors. Cells containing the desired receptors
and efficient nucleic accumulation. Other DNA devices have showed the peak activity of the luciferase that was transfected.
taken advantage of this late stage endosomal pH shift.[30] This receptor-targeting gene delivery can be apply on some
Kim et al. created a DNA nanoball composed of an RCA emphysema patients, who has deficiency of alpha1–antitrypsin
template that was complimentary and
hybridized to two types of antisense oligo-
nucleotides.[49] These were then condensed
by cationic Mu peptides and coated with
hyaluronic acid to target the CD44 recep-
tors. The addition of Mu decreased the
size and increased the stability against
nuclease.
In 2012, Lee et al. reported the con-
struction of an RNAi microsponge that
was synthesized by the self-assembly of
siRNA polymers that are cleavable after
cellular uptake, thus allowing for the pro-
tection of the siRNA until it has been
successfully delivered (Figure 6B).[73] The
RNA is stable in the pleated nanosheets
that form the microsponge as they exist
in the hairpin modality. The long stranded
RNA was produced through RCA but uti-
lizing RNA polymerase instead of DNA
polymerase. These microsponges, when
combined with cationic polyethylenimine
(PEI) were successfully transfected into
cells and showed knock-down of luciferase
expression.
Rolling circle amplification is a very Figure 7. Proposed delivery path for DNA nanoparticles. Reproduced with permission.[79]
integrative process that allows for the Copyright 2002, Springer.

small 2016, 12, No. 9, 1117–1132 © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.small-journal.com 1127
reviews
www.MaterialsViews.com

(A1AT), a specific kind of antiprotease, particularly in the 3.5. Other DNA Nanostructures
lung.[75] In this treatment, alveolar macrophages (PAM)
which reside within alveoli are targeted and mannose-ter- It is also worthwhile to review some other unique DNA nano-
minal glycoprotein conjugates are used. Although the gene structures that have been used for drug delivery that don’t
showed low expression in the cells, the gene transfer tech- fall entirely within any particular category of synthesis. These
nique still appears promising once they have found proper methods also possess the potential of shape control, ease of
receptor for this particular problem. functionalization of targeting agents, and incorporation of
This system is also used as a gene therapy for cystic drugs.
fibrosis.[78,80–83,85] Utilizing the same serpin enzyme receptor DNA blockcopolymer system was introduced by
targeting.[74,76] and condensing the plasmid DNA for the Alemdaroglu et al. in 2008 (Figure 8A).[90] The hydrophobic
complementary DNA (cDNA) encoding human cystic core made of polypropylene oxide (PPO) is designated for
fibrosis transmembrane conductance regulator (CFTR) incorporating hydrophobic drugs like doxorubicin (Figure 8A).
with poly-l-lysine, these particles can be used to correct The DNA shell makes it easier for functionalization of tar-
the chloride transfer defect.[78] These particles restored the geting groups, in this case, folic acid. The diameter of the final
expression of CFTR and of nitric oxide synthase-2 (NOS-2) particles is roughly 10 nm. Human colon adenocarcinoma
in the nasal epithelium where the complexes were admin- (Caco-2) cells were used to test this delivery system. Fluores-
istered even with only 0.15 µg of DNA administered. Sim- cence labeling showed that the particles are readily uptaken
ilar particles, with serpin targeting, that transfected the by the cells and loading of Dox showed efficient cytotoxicity.
LacZ reporter gene showed galactosidase expression but DNA can also be used to create hydrogels for targeted
no correction of the NOS-2 deficit. When these particles delivery that can be environmentally triggerable by including
are poly(ethylene glycol)-modified (PEGylated), there is aptamers and disulfide linkages.[91,92]
a higher avoidance of aggregation, higher circulation time, As Hong et al. reported, one can utilize siRNA to create
and effective transfection of the desired gene in the airway nanogels through polymeric condensation.[91] These siRNA
epithelium.[80] They also do not require the targeting ligand. modalities contained Y-branched antisense siRNA to act as a
The toxicity of these PEGylated particles is minimal in vivo, cross-linker for the short stranded sense and antisense siRNA
stable in saline, and have a half-life of 2–4 hours in mouse strand. Three modalities were synthesized: M-siRNA, which
serum.[81] These complexes also do not induce an antibody mixed the sense and antisense, DY-siRNA, which mixed the
response when lacking PEG,[84] and when tested in human sense and the antisense strand with the Y-shaped linkers, and
subjects, there were no adverse effects from the drug except YY-siRNA which combined the Y-shaped sense and anti-
in possibly one subject, and 8 out of the 12 subjects expe- sense strand (Figure 8B). The gels were condensed through
rienced at least partial, if not complete, CFTR chloride the use of cationic linear polyethylenimine (LPEI) and
channel function.[83] Repeating dosing, at least three doses without the Y-shaped linker (M-siRNA) condensed particles
have been demonstrated as possible with the targeting, were not formed. However,unlike other nanogels discussed
were also suggested as a possible to extend the lifetime of in this review, there were no acidic cleavable modalities and
the transgene expression, despite the low levels of antibody so the nanogels must be cleaved by RNA enzymes inside
formation to the ligand.[84] the cell, specifically Dicer which can cleave artificial siRNA
The PEGylated DNPs also showed long-term transgene within the cell.
expression in central nervous system in mice.[87] An intracer- Cho et al. reported the construction of DNA structured
ebral injection of DNPs can transfect both neuron and glia, nanofilm with controllable release properties.[93] Three types
and the transgene expression stayed active in the striatum of film were assembled from hairpin shaped, Y-shaped, and
for up to 8 weeks, at least 100-fold greater than naked DNA. X-shaped DNA that contained “sticky ends” and each film was
Bioluminescent imaging (BLI) was also used 11 weeks after assembled, layer-by-layer, through the use of poly-l-Lysine
injection and significant higher transgene activity by DNPs (Figure 8C). Each layer of DNA film contained Dox as an
than naked DNA was detected. intercalating agent and demonstrated different release pro-
Other than treatment, these particles are also useful files, in serum, of the drug depended on the shape of the
in real-time imaging system: BLI, positron emission DNA used. X-DNA films showed a low burst initial release
tomography (PET), and magnetic resonance imaging with a slower and more controlled release than the other two
(MRI).[86] The efficacy of gene delivery using poly(ethylene films, H-DNA had a larger initial burst release and a faster
glycol) (PEG)-stabilized DNPs was assessed using BLI of release overall. Thus, this modality can control the release of
transgene expression in wild type and cystic fibrosis mouse the doxorubicin based on the type of film.
models[88] and in long-term tracking of transgene activity in Y shaped aptamers can also be used to for big multilay-
brain in 2011.[89] ered DNA dendrimers (Figure 8D).[94] By designing each
Another kind of DNA nanoparticle includes the combi- layer of aptamers differently, the DNA dendrimers contain
nation of interfering RNA with actual metallic nanoparticles fluorophores, targeting ligands (sgc8) and anticancer drugs
to increase the density and the uptake of the siRNA.[15] (Dox) for imaging, cell recognition and drug delivery. This
These particles, reported by Jensen et al., showed significant complex also showed potent toxicity towards target cancer
down-regulation of their target: the oncoprotein, Bcl2Like12 cells (human T cell acute lymphoblastic leukemia cell line)
which inhibits apoptosis, thus allowing for higher cytotoxicity while having low side effects for the non-target cells (human
of the cancer cells. Burkitt’s lymphoma cell line).

1128 www.small-journal.com © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim small 2016, 12, No. 9, 1117–1132
www.MaterialsViews.com

Figure 8. A) DNA blockcopolymer system. Reproduced with permission.[90] Copyright 2008, Wiley-VCH. B) DNA nanogel. Reproduced with
permission.[91] Copyright 2011, American Chemical Society. C) DNA-structured nanofilm synthesized by layer by layer deposition. Reproduced with
permission.[93] Copyright 2014, Nature Publishing Group. D) DNA dendrimer layered particles. Reproduced with permission.[94] Copyright 2015,
Nature Publishing Group.

4. Summary and Future Outlook variety of sizes and shapes have been fabricated either
through oligonucleotide, RCA or origami methods, along
DNA nanotechnology has shown great potential in con- with other hybrid nanostructures. Their advantages and dis-
trolling drug/gene delivery. DNA based particles of a vast advantages are summarized in Table 1. They can be readily

small 2016, 12, No. 9, 1117–1132 © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.small-journal.com 1129
reviews
www.MaterialsViews.com
Table 1. Comparison of different DNA nanotechnology approaches. a naturally biodegradable and biocompatible
polymer, DNA shows quite promising perfor-
Approach Advantage Disadvantage mance in certain types of cells and in mouse,[52]
ODN Commercially available short strands; Less structurally stable than origami; In these early studies, there was no antibody
less types of strands involved in no available programmable designing response against DNA nanostructures. However,
structure construction. tool. given the complexity of human body, the influ-
DNA origami Programmable tools (e.g., caDNAno); Safety concern about the virus DNA ence of particle physiochemical properties on
More condensed for intercalation of backbone; hundreds of types of staple renal systems, and the unlikely but essentially
drugs. strands. harmful genome recombination, further inves-
RCA Economic and easy large-scale Lack of precise control in length and tigation on DNA nanostructures in different
fabrication. structure. types of organs are needed for their clinic appli-
DNA nanoparticles Integration of different materials Safety issue regarding combinations cation translation. iii) Undetermined large scale
properties (e.g., polymer and DNA). of materials. production. Compared with polymer and lipid
delivery systems, DNA nanostructures have
functionalized to target specific cells, elongate circulation added cost, given the synthesis challenges. A few methods,
time, respond to environmental cues, and incorporate drug, including PCR, RCA and in-cell production,[95] are used to
gene and imaging agents. Therefore, as demonstrated in this amplify the DNA strands using relatively cheap costs. How-
review, DNA nanotechnology has become a robust platform ever, considering the complexity of DNA nanostructures,
to bring controls in a lot of different aspects into drug and there is still a gap that needs to be filled. To enlarge the
gene delivery. DNA can also be easily functionalized, as production scale, besides what discussed in this review, new
discussed earlier in this review, which allows for the combi- approaches to synthesis DNA strands and DNA nanostruc-
nation of DNA nanostructures with other nanostructures. tures are also needed. We believe that with the further devel-
This can lead to creative solutions for therapeutics such as opment in drug delivery and resolution of scalability issues,
the combination DNA/gold nanoparticle therapy of Jensen DNA nanotechnology can bring a new concept into carrier
et al.[15] DNA can also be situated onto structures that are not systems and generate useful clinic results.
metallic, such as polymeric structures allowing for codelivery.
The examples we discussed demonstrate the wide breadth
of how DNA can be used to create versatile, precision nano-
structures for controllable, targeted delivery of molecular
Acknowledgements
therapeutics and gene therapeutics.
While DNA nanotechnology holds high promise for
C.A. and S.X. contributed equally to this work. This work is sup-
precision nanomedicine, it is still in very early stage. So far
ported by University of California-San Diego (faculty start-up fund).
to the authors’ knowledge, there is no significant clinical
trial in progress regarding DNA nanotechnology. Some key
issues need to be considered prior to its clinical translation.
[1] R. Langer, Science 1990, 249, 1527.
i) Unclear working mechanism. Although DNA nanostruc- [2] R. Langer, Nature 1998, 392, 5.
tures have been tested in drug delivery, further exploration [3] L. Steidler, W. Hans, L. Schotte, S. Neirynck, F. Obermeier, W. Falk,
into the mechanisms of transfection is urgently needed as the W. Fiers, E. Remaut, Science 2000, 289, 1352.
actual mechanism of uptake and how do important factors [4] C.-M. J. Hu, L. Zhang, S. Aryal, C. Cheung, R. H. Fang, L. Zhang,
like size and shape affect the uptake are still not fully under- Proc. Natl. Acad. Sci. USA 2011, 108, 10980.
stood. As these mechanisms are explored, more targeting [5] P. L. Felgner, G. M. Ringold, Nature 1989, 337, 387.
[6] V. P. Torchilin, Nat. Rev. Drug Discovery 2005, 4, 145.
modalities will become available and will allow for a higher
[7] F. Alexis, E. Pridgen, L. K. Molnar, O. C. Farokhzad, Mol.
uptake of the DNA nanostructures thus allowing for a lower Pharmaceutics 2008, 5, 505.
dosage of therapeutics and a lower possible chance of detri- [8] T. Sun, Y. S. Zhang, B. Pang, D. C. Hyun, M. Yang, Y. Xia, Angew.
mental off-target effects. As a novel delivery platform, most Chem. Int Ed. 2014, 53, 12320.
of the current reports on drug delivery applications are using [9] O. C. Farokhzad, R. Langer, ACS Nano 2009, 3, 16.
Dox as a model drug, given the easy intercalation process [10] D. Peer, J. M. Karp, S. Hong, O. C. Farokhzad, R. Margalit,
and the fluorescence property when released. To broaden R. Langer, Nat. Nanotechnol. 2007, 2, 751.
[11] Y. Geng, P. Dalhaimer, S. Cai, R. Tsai, M. Tewari, T. Minko,
their applications, it is vital to investigate the use of DNA
D. E. Discher, Nat. Nanotechnol. 2007, 2, 249.
nanotechnology on other drug systems to expand the horizon [12] J. A. Champion, S. Mitragotri, Proc. Natl. Acad. Sci. USA 2006,
of the application. Another important issue is the controlled 103, 4930.
release. Although it has been reported that the release profile [13] K.-R. Kim, D.-R. Kim, T. Lee, J. Y. Yhee, B.-S. Kim, I. C. Kwon,
can be tuned in DNA origami,[28] it is still important to con- D.-R. Ahn, Chem. Commun. 2013, 49, 2010.
trol the release profile to make a sustainable drug concen- [14] N. C. Seeman, N. R. Kallenbach, Biophys. J. 1983, 44, 201.
tration for certain treatment purpose, some of which should [15] S. A. Jensen, E. S. Day, C. H. Ko, L. A. Hurley, J. P. Luciano,
F. M. Kouri, T. J. Merkel, A. J. Luthi, P. C. Patel, J. I. Cutler,
be based on the incorporation of multiple types of drugs. For W. L. Daniel, A. W. Scott, M. W. Rotz, T. J. Meade, D. A. Giljohann,
this purpose, incorporation of multiple release mechanisms, C. A. Mirkin, A. H. Stegh, Sci. Transl. Med. 2013, 5, 209ra152.
e.g., environmental stimuli and slow degradation of the struc- [16] N. C. Seeman, Biochemistry 2003, 42, 7259.
ture can be explored. ii) Further testing of safety profile. As [17] J. Chen, N. C. Seeman, Nature 1991, 350, 631.

1130 www.small-journal.com © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim small 2016, 12, No. 9, 1117–1132
www.MaterialsViews.com
[18] R. P. Goodman, I. a. T. Schaap, C. F. Tardin, C. M. Erben, R. M. Berry, [54] C. Wu, D. Han, T. Chen, L. Peng, G. Zhu, M. You, L. Qiu, K. Sefah,
C. F. Schmidt, A. J. Turberfield, Science 2005, 310, 1661. X. Zhang, W. Tan, J. Am. Chem. Soc. 2013, 135, 18644.
[19] Y. He, T. Ye, M. Su, C. Zhang, A. E. Ribbe, W. Jiang, C. Mao, Nature [55] J. Li, H. Pei, B. Zhu, L. Liang, M. Wei, Y. He, N. Chen, D. Li,
2008, 452, 198. Q. Huang, C. Fan, ACS Nano 2011, 5, 8783.
[20] D. M. Smith, V. Schüller, C. Forthmann, R. Schreiber, P. Tinnefeld, [56] E. S. Andersen, M. Dong, M. M. Nielsen, K. Jahn, A. Lind-Thomsen,
T. Liedl, J. Nucleic Acids 2011, 2011, e360954. W. Mamdouh, K. V. Gothelf, F. Besenbacher, J. Kjems, ACS Nano
[21] Y. Zhang, N. C. Seeman, J. Am. Chem. Soc. 1994, 116, 1661. 2008, 2, 1213.
[22] C. Zhang, M. Su, Y. He, X. Zhao, P.-a. Fang, A. E. Ribbe, W. Jiang, [57] A. E. Marras, L. Zhou, H.-J. Su, C. E. Castro, Proc. Natl. Acad. Sci.
C. Mao, Proc. Natl. Acad. Sci. USA 2008, 105, 10665. USA 2015, 112, 713.
[23] D. Bhatia, S. Mehtab, R. Krishnan, S. S. Indi, A. Basu, Y. Krishnan, [58] P. W. K. Rothemund, Nature 2006, 440, 297.
Angew. Chem. Int Ed. 2009, 48, 4134. [59] T. Tørring, N. V. Voigt, J. Nangreave, H. Yan, K. V. Gothelf, Chem.
[24] R. Iinuma, Y. Ke, R. Jungmann, T. Schlichthaerle, J. B. Woehrstein, Soc. Rev. 2011, 40, 5636.
P. Yin, Science 2014, 344, 65. [60] S. M. Douglas, H. Dietz, T. Liedl, B. Högberg, F. Graf, W. M. Shih,
[25] Y. He, Y. Chen, H. Liu, A. E. Ribbe, C. Mao, J. Am. Chem. Soc. 2005, Nature 2009, 459, 414.
127, 12202. [61] S. M. Douglas, A. H. Marblestone, S. Teerapittayanon, A. Vazquez,
[26] M. Chang, C.-S. Yang, D.-M. Huang, ACS Nano 2011, 5, 6156. G. M. Church, W. M. Shih, Nucleic Acids Res. 2009, gkp436.
[27] Y. Ke, L. L. Ong, W. M. Shih, P. Yin, Science 2012, 338, 1177. [62] Q. Zhang, Q. Jiang, N. Li, L. Dai, Q. Liu, L. Song, J. Wang, Y. Li,
[28] Y.-X. Zhao, A. Shaw, X. Zeng, E. Benson, A. M. Nyström, J. Tian, B. Ding, Y. Du, ACS Nano 2014, 8, 6633.
B. Högberg, ACS Nano 2012, 6, 8684. [63] W. M. Shih, J. D. Quispe, G. F. Joyce, Nature 2004, 427, 618.
[29] W. Sun, T. Jiang, Y. Lu, M. Reiff, R. Mo, Z. Gu, J. Am. Chem. Soc. [64] J.-P. J. Sobczak, T. G. Martin, T. Gerling, H. Dietz, Science 2012,
2014, 136, 14722. 338, 1458.
[30] S. Surana, J. M. Bhat, S. P. Koushika, Y. Krishnan, Nat. Commun. [65] X. Huang, C. S. Brazel, J. Controlled Release 2001, 73, 121.
2011, 2, 340. [66] Q. Jiang, C. Song, J. Nangreave, X. Liu, L. Lin, D. Qiu, Z.-G. Wang,
[31] D. Agudelo, P. Bourassa, G. Bérubé, H.-A. Tajmir-Riahi, Int. J. Biol. G. Zou, X. Liang, H. Yan, B. Ding, J. Am. Chem. Soc. 2012, 134,
Macromol. 2014, 66, 144. 13396.
[32] G. Zhu, S. Cansiz, M. You, L. Qiu, D. Han, L. Zhang, L. Mei, T. Fu, [67] S. M. Douglas, I. Bachelet, G. M. Church, Science 2012, 335, 831.
Z. Chen, W. Tan, NPG Asia Mater. 2015, 7, e169. [68] Y. Mao, M. Liu, K. Tram, J. Gu, B. J. Salena, Y. Jiang, Y. Li, Chem.
[33] C. Fellmann, S. W. Lowe, Nat. Cell Biol. 2014, 16, 10. Eur. J. 2015, 21, 8069.
[34] Y. Naito, K. Ui-Tei, RNA 2012, 3, 102. [69] X. Ouyang, J. Li, H. Liu, B. Zhao, J. Yan, Y. Ma, S. Xiao, S. Song,
[35] D. Castanotto, J. J. Rossi, Nature 2009, 457, 426. Q. Huang, J. Chao, C. Fan, Small 2013, 9, 3082.
[36] P. Guo, O. Coban, N. M. Snead, J. Trebley, S. Hoeprich, S. Guo, [70] G. D. Hamblin, K. M. M. Carneiro, J. F. Fakhoury, K. E. Bujold,
Y. Shu, Adv. Drug. Deliv. Rev. 2010, 62, 650. H. F. Sleiman, J. Am. Chem. Soc. 2012, 134, 2888.
[37] L. Liu, K. Xu, H. Wang, P. K. J. Tan, W. Fan, S. S. Venkatraman, L. Li, [71] J. Yan, C. Hu, P. Wang, B. Zhao, X. Ouyang, J. Zhou, R. Liu, D. He,
Y.-Y. Yang, Nat. Nanotechnol. 2009, 4, 457. C. Fan, S. Song, Angew. Chem. 2015, 127, 2461.
[38] O. I. Wilner, I. Willner, Chem. Rev. 2012, 112, 2528. [72] W. Xu, R. Deng, L. Wang, J. Li, Anal. Chem. 2014, 86, 7813.
[39] T. M. Allen, Nat. Rev. Cancer 2002, 2, 750. [73] J. B. Lee, J. Hong, D. K. Bonner, Z. Poon, P. T. Hammond, Nat.
[40] E. Cheng, Y. Xing, P. Chen, Y. Yang, Y. Sun, D. Zhou, L. Xu, Q. Fan, Mater. 2012, 11, 316.
D. Liu, Angew. Chem. 2009, 121, 7796. [74] A. G. Ziady, J. C. Perales, T. Ferkol, T. Gerken, H. Beegen,
[41] S. Modi, M. G. Swetha, D. Goswami, G. D. Gupta, S. Mayor, D. H. Perlmutter, P. B. Davis, Am. J. Physiol. 1997, 273, G545.
Y. Krishnan, Nat. Nanotechnol. 2009, 4, 325. [75] T. Ferkol, F. Mularo, J. Hilliard, S. Lodish, J. C. Perales, A. Ziady,
[42] T. Lühmann, M. Rimann, A. G. Bittermann, H. Hall, Bioconjugate M. Konstan, Am. J. Respir. Cell Mol. Biol. 1998, 18, 591.
Chem. 2008, 19, 1907. [76] A. Ziady, T. Ferkol, T. Gerken, D. Dawson, D. Perlmutter, P. Davis,
[43] G. Chen, D. Liu, C. He, T. R. Gannett, W. Lin, Y. Weizmann, J. Am. Gene Ther. 1998, 5, 1685.
Chem. Soc. 2015, 137, 3844. [77] A.-G. Ziady, T. Ferkol, D. V. Dawson, D. H. Perlmutter, P. B. Davis,
[44] S. Xiang, H. Tong, Q. Shi, J. C. Fernandes, T. Jin, K. Dai, X. Zhang, J. Biol. Chem. 1999, 274, 4908.
J. Controlled Release 2012, 158, 371. [78] A.-G. Ziady, T. J. Kelley, E. Milliken, T. Ferkol, P. B. Davis, Mol. Ther.
[45] X. Chen, S. Shank, P. B. Davis, A. G. Ziady, Mol. Ther. 2011, 19, 2002, 5, 413.
93. [79] A. Ziady, P. Davis, in Gene Therapy Protocols (Ed: J. Morgan),
[46] B. Dalby, S. Cates, A. Harris, E. C. Ohki, M. L. Tilkins, P. J. Price, Vol. 69, Springer, New York 2002, pp. 25.
V. C. Ciccarone, Methods 2004, 33, 95. [80] A.-G. Ziady, C. R. Gedeon, T. Miller, W. Quan, J. M. Payne,
[47] Y. Ke, G. Bellot, N. V. Voigt, E. Fradkov, W. M. Shih, Chem. Sci. S. L. Hyatt, T. L. Fink, O. Muhammad, S. Oette, T. Kowalczyk,
2012, 3, 2587. M. K. Pasumarthy, R. C. Moen, M. J. Cooper, P. B. Davis, Mol. Ther.
[48] J. Mikkilä, A.-P. Eskelinen, E. H. Niemelä, V. Linko, M. J. Frilander, 2003, 8, 936.
P. Törmä, M. A. Kostiainen, Nano Lett. 2014, 14, 2196. [81] A.-G. Ziady, C. R. Gedeon, O. Muhammad, V. Stillwell, S. M. Oette,
[49] M.-G. Kim, J. Y. Park, G. Shim, H.-G. Choi, Y.-K. Oh, Biomaterials T. L. Fink, W. Quan, T. H. Kowalczyk, S. L. Hyatt, J. Payne,
2015, 62, 155. A. Peischl, J. E. Seng, R. C. Moen, M. J. Cooper, P. B. Davis, Mol.
[50] S. Sellner, S. Kocabey, K. Nekolla, F. Krombach, T. Liedl, Ther. 2003, 8, 948.
M. Rehberg, Biomaterials 2015, 53, 453. [82] A. G. Ziady, P. B. Davis, M. W. Konstan, Expert Opin. Biol. Ther.
[51] R. P. Goodman, R. M. Berry, A. J. Turberfield, Chem. Commun. 2003, 3, 449.
2004, 1372. [83] M. W. Konstan, P. B. Davis, J. S. Wagener, K. A. Hilliard, R. C. Stern,
[52] H. Lee, A. K. R. Lytton-Jean, Y. Chen, K. T. Love, A. I. Park, L. J. H. Milgram, T. H. Kowalczyk, S. L. Hyatt, T. L. Fink, C. R. Gedeon,
E. D. Karagiannis, A. Sehgal, W. Querbes, C. S. Zurenko, S. M. Oette, J. M. Payne, O. Muhammad, A. G. Ziady, R. C. Moen,
M. Jayaraman, C. G. Peng, K. Charisse, A. Borodovsky, M. J. Cooper, Human Gene Ther. 2004, 15, 1255.
M. Manoharan, J. S. Donahoe, J. Truelove, M. Nahrendorf, [84] A.-G. Ziady, J. Kim, J. Colla, P. B. Davis, Gene Ther. 2004, 11, 1378.
R. Langer, D. G. Anderson, Nat. Nanotechnol. 2012, 7, 389. [85] A. G. Ziady, P. B. Davis, Curr. Opin. Pharmacol. 2006, 6, 515.
[53] G. Zhu, J. Zheng, E. Song, M. Donovan, K. Zhang, C. Liu, W. Tan, [86] W. Sun, A. G. Ziady, in Micro and Nano Technologies in Bioanalysis,
Proc. Natl. Acad. Sci. USA 2013, 110, 7998. Springer, New York 2009, pp. 525.

small 2016, 12, No. 9, 1117–1132 © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.small-journal.com 1131
reviews
www.MaterialsViews.com
[87] D. M. Yurek, A. M. Fletcher, G. M. Smith, K. B. Seroogy, A. G. Ziady, [92] J. Li, C. Zheng, S. Cansiz, C. Wu, J. Xu, C. Cui, Y. Liu, W. Hou, Y. Wang,
J. Molter, T. H. Kowalczyk, L. Padegimas, M. J. Cooper, Mol. Ther. L. Zhang, I.-t. Teng, H.-H. Yang, W. Tan, J. Am. Chem. Soc. 2015,
2009, 17, 641. 137, 1412.
[88] A. G. Ziady, M. Kotlarchyk, L. Bryant, M. McShane, Z. Lee, Microsc. [93] Y. Cho, J. B. Lee, J. Hong, Sci. Rep. 2014, DOI: 10.1038/srep04078.
Res. Tech. 2010, 73, 918. [94] H. Zhang, Y. Ma, Y. Xie, Y. An, Y. Huang, Z. Zhu, C. J. Yang, Sci. Rep.
[89] D. M. Yurek, A. M. Fletcher, M. McShane, T. H. Kowalczyk, 2015, DOI: 10.1038/srep10099.
L. Padegimas, M. R. Weatherspoon, M. D. Kaytor, M. J. Cooper, [95] C. Lin, S. Rinker, X. Wang, Y. Liu, N. C. Seeman, H. Yan, Proc. Natl.
A. G. Ziady, Mol. Imaging 2011, 10, 327. Acad. Sci. USA 2008, DOI: 10.1073/pnas.0805416105.
[90] F. E. Alemdaroglu, N. C. Alemdaroglu, P. Langguth, A. Herrmann,
Adv. Mater. 2008, 20, 899. Received: July 21, 2015
[91] C. A. Hong, S. H. Lee, J. S. Kim, J. W. Park, K. H. Bae, H. Mok, Revised: September 3, 2015
T. G. Park, H. Lee, J. Am. Chem. Soc. 2011, 133, 13914. Published online: January 3, 2016

1132 www.small-journal.com © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim small 2016, 12, No. 9, 1117–1132

You might also like